A method of protecting a warm-blooded mammal from the progression of Alzheimer's disease, which comprises administering an effective amount of a compound of general formula. ##STR1## in which: R1 represents an optionally substituted aromatic or heteroaromatic group;

L1 and L2 each independently represents a bond or an unbranched (1-4C) alkylene group, which alkylene group may optionally bear a (1-4C) alkyl, phenyl or phenyl (1-2C) alkyl substituent;

one of Y1 and Y2 represents NR3 and the other represents O, S or NR3, in which R3 represent hydrogen, hydroxyl, (1-4C) alkoxy, (1-4C) alkyl or di (1-4C) alkylamino;

Z represents O or S; and

R2 represents (3-8C) cycloalkyl, heterocyclyl, and optionally substituted aromatic or heteroaromatic group, or together with L2, (1-10C) alkyl;

or a pharmaceutically acceptable salt thereof.

Patent
   5814646
Priority
Mar 02 1995
Filed
Mar 02 1995
Issued
Sep 29 1998
Expiry
Sep 29 2015
Assg.orig
Entity
Large
38
13
EXPIRED
1. A method of inhibiting the production of Aβ peptide in a biological system, which comprises administering an effective amount of a compound of general formula ##STR7## in which Z is O or S;
R11 is a halogen atom;
R12 is a halogen atom or a trifluoromethyl group; and
R7 is hydrogen, (1-8C)alkyl, (1-8C)haloalkyl, (1-8C) alkylthio, (1-8C)alkoxy or halogen;
or a pharmaceutically acceptable salt thereof.
5. A method of inhibiting the accumulation of Aβ peptide in senile plaques in a warm-blooded mammal, which comprises administering an effective amount of a compound of general formula. ##STR8## in which Z is O or S;
R11 is a halogen atom;
R12 is a halogen atom or a trifluoromethyl group; and
R7 is hydrogen, (1-8C)alkyl, (1-8C)haloalkyl, (1-8C)alkylthio, (1-8C)alkoxy or halogen;
or a pharmaceutically acceptable salt thereof.
6. A method of protecting a warm-blooded mammal from the progression of Alzheimer's disease by inhibiting the formation of Aβ peptide, which comprises administering an effective amount of a compound of general formula. ##STR9## in which Z is O or S;
R11 is a halogen atom;
R12 is a halogen atom or a trifluoromethyl group; and
R7 is hydrogen, (1-8C)alkyl, (1-8C)haloalkyl, (1-8C) alkylthio, (1-8C)alkoxy or halogen;
or a pharmaceutically acceptable salt thereof.
2. A method as claimed in claim 1, in which Z is O, R11 is a fluorine or chlorine atom, and R12 is a chlorine atoms or a trifluoromethyl group.
3. A method as claimed in claim 2 in which R7 is hydrogen, methyl, t-butyl, trifluoromethyl, 2chloro-2-methylpropyl, hexylthio, bromo or chloro.
4. A method as claimed in claim 1, in which the compound of formula 5 is selected from 1-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;
1-(5-chloro-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;
1-(5-trifluoromethyl-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;
1-(5-t-butyl-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;
1-[5-(2-chloro-1,1-dimethylethyl)-1,3,4-thiadiazol-2-yl]-3-(3,4-dichlorophe nyl)urea;
1-(5-hexylthio-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;
1-(5-bromo-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;
1-(5-methyl-1,3,4-thiadiazolyl)-3-(4-fluoro-3-chlorophenyl)urea; and
1-(5-methyl-1,3,4-thiadiazolyl)-3-(3,4-dichlorophenyl)urea or a pharmaceutically acceptable salt thereof.

The present invention relates to the use of certain urea derivatives as inhibitors of amyloid beta-protein production. It also relates to novel urea derivatives, to processes for their preparation and to pharmaceutical compositions containing them.

Amyloid beta-protein (Aβ) is a neurotoxic polypeptide containing about 40 amino acid residues. It is produced by enzymatic cleavage of a larger precursor protein, beta-amyloid precursor protein, which is encoded by a gene on human chromosome 21, and is found in the brains of individuals suffering from Alzheimer's disease in deposits known as senile plaques. It is now widely believed that Aβ is involved in the pathogenesis of Alzheimer's disease, and substantial efforts are being made to find ways of intervening in this involvement, for example by inhibiting the production of Aβ.

It has now been found that certain urea derivatives, some of which are novel, are capable of inhibiting the production of Aβ in cells.

The present invention provides a method of inhibiting the production of Aβ peptide in a biological system, which comprises administering an effective amount of a compound of general formula. ##STR2## in which: R1 represents an optionally substituted aromatic or heteroaromatic group;

L1 and L2 each independently represents a bond or an unbranched (1-4C) alkylene group, which alkylene group may

optionally bear a (1-4C) alkyl, phenyl or phenyl (1-2C) alkyl substituent;

one of Y1 and Y2 represents NR3 and the other represents O, S or NR3, in which R3 represent hydrogen, hydroxyl, (1-4C) alkoxy, (1-4C) alkyl or di (1-4C) alkylamino;

Z represents O or S; and

R2 represents (3-8C) cycloalkyl, heterocyclyl, an optionally substituted aromatic or heteroaromatic group, or together with L2, (1-10C) alkyl;

or a pharmaceutically acceptable salt thereof.

It has been found that urea derivatives of formula I are capable of inhibiting the production of Aβ in whole cells. Accordingly, it is believed that these compounds will be capable of inhibiting the production of Aβ generally in biological systems, and will be capable of inhibiting the accumulation of Aβ in senile plaques in a warm blooded mammal, such as man. The urea derivatives should therefore be capable of protecting a warm blooded mammal, such as man, from the progression of Alzheimer's disease.

According to another aspect, therefore, the present invention provides a method of inhibiting the accumulation of Aβ in senile plaques in a warm blooded mammal, which comprises administering an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.

According to yet another aspect, the present invention provides a method of protecting a warm blooded mammal from the progression of Alzheimer's disease, which comprises administering an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.

The present invention also provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of Alzheimer's disease.

The urea derivatives which have been found to be capable of inhibiting the production of Aβ in whole cells possess a diverse range of structures. Accordingly it is believed that they are representatives of a new, broad structural class of inhibitor of Aβ production, as defined herein with reference to formula I.

As used herein with reference to formula I, the term aromatic group includes phenyl and naphthyl.

The term heteroaromatic group includes a5-6 membered ring containing a heteroatom selected from oxygen, sulfur and nitrogen and up to three additional nitrogen atoms, the remaining ring atoms all being carbon atoms, said ring, optionally being fused to a benzene ring. Examples of heteroaromatic groups include furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, imidazolyl, pyridyl, pyrimidyl, triazinyl, benzofuryl, benzothienyl, benzothiazolyl, benzimidazolyl, indolyl and quinolyl.

The term optionally substituted, as used in optionally substituted aromatic or heteroaromatic group means that the group is unsubstituted or substituted by one or more, for example 1, 2 or 3, substituents which do not interfere with the ability of the compound of formula I to inhibit Aβ production.

Examples of substituents which may be present in an optionally substituted aromatic or heteroaromatic group (for example an optionally substituted phenyl group) include hydroxy, halogeno, (1-8C) alkyl, methylenedioxy, carboxy, (1-8C) alkoxycarbonyl, halo(1-8C) alkyl and groups of formula XR4, in which R4 represents (1-6C) alkyl, phenyl which is unsubstituted or substituted by 1,2 or 3 of (1-6C) alkyl, (1-6C) alkoxy, amino, halogeno, (1-6C) alkanoyl, benzoyl, phenyl, hydroxy and halo (1-6C) alkyl, naphthyl, furyl, thienyl, pyridyl, benzothienyl, quinolyl, indolyl and benzimidazolyl, and X represents O, CH2, OCH2, S, SO, SO2, NH, CO or CONH.

The term (1-8C) alkyl includes (1-6C) alkyl and (1-4C) alkyl. Examples of (1-8C) alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl and hexyl.

The term (1-8C) alkoxy includes (1-6C) alkoxy and (1-4C) alkoxy. Examples of a (1-8C) alkoxy group are methoxy, ethoxy and butoxy.

Examples of a halogen atom are fluoro, chloro and bromo.

Halo(1-8C) alkyl includes halo (1-6C) alkyl, for example trifluoromethyl.

An example of a (1-8C) alkoxycarbonyl group is methoxycarbonyl.

An example of a (1-6C) alkanoyl group is acetyl.

Examples of a (1-4C) alkylene group are methylene and ethylene.

An example of a phenyl(1-2C)alkyl group is benzyl.

An example of a di (1-4C) alkylamino group is dimethylamino.

Examples of a (3-8C) cycloalkyl group are cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.

The term heterocyclyl, as used herein includes a non-aromatic 4-6 membered ring containing one or two heteroatoms selected independently from oxygen, sulfur and nitrogen, the remaining ring atoms all being carbon atoms. An example of heterocyclyl is piperidinyl.

Preferably R2 represents cyclopentyl; cyclohexyl; piperdinyl; phenyl which is unsubstituted or substituted by methylenedioxy or by one or two substituents independently selected from halogeno, (1-4C) alkyl, (1-4C) alkoxy, fluoro (1-4C) alkyl and fluoro (1-4C) alkoxy; naphthyl; pyridyl or, together with L2, represents 5-10C) alkyl.

More preferably R2 represents a phenyl group which is unsubstituted or substituted by one or two substituents selected from fluoro, chloro, methyl, methoxy and trifluoromethyl.

Examples of specific values for R2 are 2,3-dichlorophenyl, 4,5-dichloro-2-methylphenyl, 4-methylthiophenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-t-butylphenyl, 2,6-dimethylphenyl, 3-methylthiophenyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 4-fluoromethyl, 4-chlorophenyl, 3,4-dichlorophenyl, 3-chloro -4-methylphenyl, 3,4-dimethylphenyl, 3,4-dimethoxyphenyl, 3,4-methylenedioxyphenyl, 3-chlorophenyl, 2,4-dichlorophenyl, 3-fluorophenyl, 3-methoxyphenyl, 2-chloro-3-trifluoro-methylphenyl, 4-chloro-3-trifluoromethylphenyl, 4-(n -butoxy)phenyl, 2-chlorophenyl, 3-bromophenyl, 4-bromophenyl, 2-methoxyphenyl, 4-methoxyphenyl, 2,4-difluorophenyl, 3,4-difluorophenyl, 2-chloro-4-fluorophenyl, 3-chloro-4-fluorophenyl, 4-fluoro-2-trifluoromethylphenyl, 3,4,5-trichlorophenyl, 1-naphthyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, N-t-butoxycarbonyl-2-piperidinyl, N-t-butoxycarbonyl-3-piperidinyl, 2-piperidinyl, 3-piperidinyl and 2-trifluoromethyl-1,3,4-thiadiazoyl, or together with L2, methyl or hexyl.

Most preferably R2 represents 3,4-dichlorophenyl or 4-chloro -3-trifluoromethylphenyl.

R1 preferably represents a phenyl, naphthyl, furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, imidazolyl, pyridyl, pyrimidyl, triazinyl, benzofuryl, benzothienyl, benzothiazolyl or benzimidazolyl group, which group is unsubstituted or substituted by one or two substituents independently selected from hydroxy, halogeno, (1-6C) alkyl, carboxy, (1-4C) alkoxycarbonyl, halo (1-6C) alkyl and groups of formula XR4 in which R4 represents (1-6C)alkyl, phenyl which is unsubstituted or substituted by one, two or three of (1-6C) alkyl, (1-6C) alkoxy, amino, halogeno, (1-6C) alkanoyl, benzoyl, phenyl, hydroxy and halo (1-6C) alkyl, naphthyl, furyl, thienyl, pyridyl, benzothienyl, quinolyl, indolyl and benzimidazolyl and X represents O, CH2, OCH2, S, SO, SO2, NH, CO or CONH.

More preferably R1 represents a 3-phenyl, 4-phenyl or 1,3,4-thiadiazol-2-yl group which is unsubstituted or substituted as defined hereinabove.

For example R1 may represent a group of formula IIa or IIb ##STR3## in which one of R5 and R6 represents hydrogen and the other represents a group of formula XR4, and R7 represents hydrogen, (1-8C) alkyl, halo (1-8C) alkyl, (1-8C) alkylthio, (1-8C) alkoxy or halogeno.

Examples of specific values for R1 are selected from 4-(4-(t-butyl)phenoxy)phenyl, 4-(2-quinolinylthio)phenyl, 3-phenoxyphenyl, 4-phenoxyphenyl, 3-(4-t-butylphenoxy)phenyl, 3-(4-n-butylphenoxy)phenyl, 4-(3-t-butylphenoxy)phenyl, 2-(4-t-butylphenoxy)phenyl, 3-(3-t-butylphenoxy)phenyl, 4-(2-t-butylphenoxy)phenyl, 3-(4-t -butylbenzyloxy)phenyl, 4-(4-t-butylbenzyloxy)phenyl, 4-(2-phenylbenzyloxy)phenyl), 4-(3,4-dichlorobenzyloxy)phenyl, 4-(3,4-difluorobenzyloxy)phenyl, 4-(2,5-dimethylbenzyloxy)-phenyl, 4-(3,5-dimethoxybenzyloxy)phenyl, 4-(4-n-butylbenzyloxy)phenyl, 4-(2,6-dichlorobenzyloxy)-phenyl, 3-(4-butoxybenzyloxyphenyl, 4-(4-acetyl-3-hydroxy-2-propylbenzyloxy)phenyl, 4-(4-benzoyl-3-hydroxy-2-propylbenzyloxy)phenyl, 2-propylbenzyloxy)phenyl, 3-(3,5-di-t-butyl-4-methoxybenzyloxy)phenyl, 4-(3,5-di-t-butyl-4-methoxybenzyloxy)phenyl, 2-(2-phenylbenzyloxy)phenyl, 4-(2-phenylbenzyloxy)phenyl, 4-biphenyl, 4-(4-t-butylphenyl)-phenyl, 3-(4-t-butylbenzamido)phenyl, 3-(4-t-butylphenylthio)phenyl, 4-(4-t-butylphenylthio)phenyl, 3-(4-t-butylphenylsulfinyl)phenyl, 3-(4-t-butylphenylsulfonyl)-phenyl, 4-(4-t-butylphenylthiomethyl)phenyl, 4-(4-1,1-dimethylpropylphenoxymethyl)phenyl, 3-(2-propyl-4-t-butylphenoxyphenyl, 5-quinolinyl, 2-benzothiazolyl, 2-pyrimidinyl, 4-chlorophenyl, 2-benzimidazolyl, 2-dimethylaminopropylthiophenyl, 2-methoxycarbonylphenyl, 2-(5,6-dimethyl)benzimidazolyl, 2,4-dimethoxyphenyl, 4-(3,4-dichlorophenylthio)phenyl, 3-trifluoromethylphenyl, 4-methylthiophenyl, 3-bromo-4-methylthiophenyl, 2-chlorophenyl, phenyl, 3,4-dichlorophenyl, 4-methylsulfonylphenyl, 3-bromo-4-methylphenyl, 2,5-dichloro-4-methylthiophenyl, 3-bromo-4-methylthiophenyl, 3-methoxy-4-methoxycarbonylphenyl, 4-chloro-3-trifluoromethylphenyl, 4-(4-t-butyl-2-propylphenoxy)phenyl, 4-acetyl-3-hydroxy-2-propylphenyl, 3,4-dichlorophenyl, 4-(4-(ethoxycarbonyl-2-quinolylthio)phenyl, 4-(6-chloro-2-quinolylthio)phenyl, 4-(4-phenyl-2-quinolylthio)phenyl, 4-(4-quinolylthio)phenyl, 4-(7-trifluoromethyl-4-quinolylthio)phenyl, 4-(2-naphthylthio)phenyl, 4-(1-naphthylthio)phenyl, 4-(2-benzimidazolylthio)phenyl, 4-(2-benzoxazolylthio)phenyl, 4-(2-benzothiazolylthio)phenyl, 4-(5-chloro-2-benzothiazolylthio)phenyl, 4-(2-quinolylsulfonyl)phenyl, 4-(2-quinolylsulfonyl)phenyl, 4-(2-quinolylamino)phenyl, 4-(2-quinolyloxy)phenyl, 4-(2-quinolylthio)phenyl, 4-(6-phenyl-3-pyridazylthio)phenyl, 4-(2-pyridylthio)phenyl, 4-(5-nitroquinolylthio)phenyl, 4-(4-isopropylsulfonyl)phenyl, 2-pyridyl, 6-1,2,5-triazinyl, 1-piperidinyl, 1-ethyl-3-piperdinyl, 2,2,6,6-tetramethyl-4-piperidinyl, 5-chloro-1,3,4-thiadiazol-2-yl, 2-methoxycarbonylbenzothiophen-3-yl, 5-methyl-1,3,4-thiadiazol-2-yl, 1,3,4-thiadiazol-2-yl, 5-trifluoromethyl-1,3,4-thiadiazol-2-yl, 5-t-butyl-1,3,4-thiadiazol-2-yl, 5-(2-chloro-1,1-dimethylethyl)-1,3,4-thiadiazol-2-yl, 5-hexylthio-1,3,4-thiadiazol-2-yl, 5-bromo-1,3,4-thiadiazol-2-yl, 2-oxazolyl, 5-methylisoxazol-3-yl, 2-thiazolyl, 4,5-dihydrothiazolyl, 3-t-butylisoxazol-5yl, and 3-t-butyl-1,2,4-oxadiazol-5-yl.

Preferably one of Y1 and Y2 represents NH and the other represents S, NH or NOH.

One of L1 and L2 preferably represents a bond and the other preferably represents a bond or methylene, ethylidene, propylidene, heptylidine or benzylidine. More preferably L1 and L2 each represents a bond.

A preferred group of compounds of formula I are those of the formula Ia ##STR4## in which Z is O or S;

R11 is a halogen atom;

R12 is a halogen atom or a trifluoromethyl group; and

R7 is hydrogen, (1-8C)alkyl, (1-8C)haloalkyl, (1-8C)alkylthio, (1-8C)alkoxy or halogen;

or a pharmaceutically acceptable salt thereof.

In this group of compounds Z is preferably 0, R11 is preferably a fluorine or chlorine atom, and R12 is preferably a chlorine atom or a trifluoromethyl group. Examples of values for R7 are hydrogen, methyl, t-butyl, trifluoromethyl, 2-chloro-2-methylpropyl, hexylthio, bromo and chloro.

Another preferred group of compounds of formula I are those of the formula Ib ##STR5## in which Z is O or S;

R11 is a halogen atom;

R12 is a halogen atom or a trifluoromethyl group;

X is S, SO, SO2, O or NH; and

R4 is naphthyl, quinolinyl, benzimidazolyl, pyridyl, pyridazinyl, benzoxazolyl or benzothiazolyl, unsubstituted or substituted by one or two substituents selected from a halogen atom, (1-4C)alkyl, (1-4C)alkoxy, nitro, (1-4C)alkoxycarbonyl, halo(1-4C)alkyl, and phenyl;

or a pharmaceutically acceptable salt thereof.

In this group of compounds, X is preferably S, Z is preferably S, R4 is preferably unsubstituted or substituted naphth-1-yl, naphth-2-yl, quinolin-2-yl, quinolin-4-yl, quinolin-6-yl, benzimidazol-2-yl, benzoxazol-2-yl or benzothiazol-2-yl, and R11 is preferably chlorine and R12 is preferably chlorine or trifluoromethyl. Examples of particular values for R4 are naphth-1-yl, naphth-2-yl, quinolin-2-yl, 6-chloroquinolin-2-yl, 6-methoxyquinolin-2-yl, 5-nitroquinolin-6-yl, 4-ethoxycarbonylquinolin-2-yl, 4-phenylquinolin-2-yl, 7-trifluoromethylquinolin-4-yl, quinolin-4-yl, benzimidazol-2-yl, benzoxazol-2-yl, benzothiazol-2-yl, 5-chloro-benzothiazol-2-yl, 6-phenylpyrazin-3-yl or pyridin-2-yl.

Particularly preferred compounds of formula I for use in the method according to the invention are 1-1,3,4-thiadiazol-2-yl-3-(3,4-dichlorophenyl)urea;

1-(5-chloro-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;

1-(5-trifluoromethyl-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;

1-(5-t-butyl-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;

1-[5-(2-chloro-1,1-dimethylethyl)-1,3,4-thiadiazol-2-yl]-3-(3,4-dichlorophe nyl)urea;

1-(5-hexylthio-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;

1-(5-bromo-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea;

1-(5-methyl-1,3,4-thiadiazol-2-yl)-3-(4-fluoro-3-chlorophenyl)urea; and

1-(5-methyl-1,3,4-thiadiazol-2-yl)-3-(3,4-dichlorophenyl)urea.

Some of the compounds of formula I, for example the compounds of formula Ib, are believed to be novel.

The present invention also provides the novel compounds of formula I, and their pharmaceutically acceptable salts, processes for their preparation and pharmaceutical compositions containing them.

The term "pharmaceutically acceptable salts" refers to salts of the compounds of the above formula which are substantially non-toxic to living organisms. Typical pharmaceutically acceptable salts include those salts prepared by reaction of the compounds of the above formula I with a pharmaceutically acceptable mineral or organic acid, or a pharmaceutically acceptable alkali metal or organic base, depending on the types of substituents present on the compounds of the formula.

Examples of pharmaceutically acceptable mineral acids which may be used to prepare pharmaceutically acceptable salts include hydrochloric acid, phosphoric acid, sulfuric acid, hydrobromic acid, hydriodic acid, phosphorous acid and the like. Examples of pharmaceutically acceptable organic acids which may be used to prepare pharmaceutically acceptable salts include aliphatic mono and dicarboxylic acids, oxalic acid, carbonic acid, citric acid, succinic acid, phenyl-substituted alkynic acids, aliphatic and aromatic sulfonic acids and the like. Such pharmaceutically acceptable salts prepared from mineral or organic acids thus include hydrochloride, hydrobromide, nitrate, sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, hydroxide, hydrofluoride, acetate, propionate, formate, oxalate, citrate, lactate, p-toluenesulfonate, methanesulfonate, maleate, and the like.

The compounds of formula I may be prepared by methods well known in the art, many compounds of formula I being known. See for example, U.S. Pat. No. 3,990,879.

Thus, compounds of formula I in which Z is O or S may be prepared by reacting a compound of formula III

R1 --L1 --A1 III

with a compound of formula IV

A2 --L2 --R2 IV

in which one of A1 and A2 represents --NH2 and the other represents --NCZ or --NHC(Z)Za in which Z is O or S, and Za is a leaving atom or group.

Za may represent, for example, an amine group such as --NH2, an alkythio group such as methylthio an aryloxy group such as phenoxy, or an acetylene group, such as a (2-10C) alk-1-ynyl group.

When the other of A1 and A2 represents --NCZ, the reaction is conveniently performed in the presence of solvent such as water, an ether, for example tetrahydrofuran, a halogenated hydrocarbon, such as dichloromethane, or an amide such as dimethylformamide. The temperature at which the reaction is conducted is conveniently in the range of from 0° to 150°C Optionally the reaction may be performed in the presence of a base, for example a tertiary amine, such as dimethylaminopyridine, or an alkali metal carbonate such as potassium carbonate.

When the other of A1 and A2 represent --NHC(Z)Za in which Za represents an acetylene group, the reaction is preferably performed in the presence of an alkali metal amide, such as sodium amide. The reaction is conveniently performed at a temperature in the range of from 0° to 50°C

When the other of A1 and A2 represents --NHC(Z)Za in which Za represents a leaving atom or group other than an acetylene group, the reaction is conveniently performed at a temperature in the range of from 0° to 150°C Suitable solvents include acetic acid, esters, such as ethyl acetate and ethers such as tetahydrofuran.

The compounds of formula III and IV in which the other of A1 and A2 represents --NC or --NCH(Z)Za may be prepared by methods known in the art from the corresponding compounds of formula III or IV respectively in which A1 or A2 respectively represents --NH2. For example, a compound of formula III or IV in which A1 or A2 represents --NH2 may be converted into the corresponding compound in which A1 or A2 represents --NCZ by reaction with phosgene, and into a corresponding compound in which A1 or A2 represents --NHCONH2 by reaction with an alkali metal cyanate, such as sodium cyanate.

The ability of a compound to inhibit the production of Aβ in a biological system may be demonstrated by the following test method.

Two cell lines (human kidney cell line 293 and Chinese hamster ovary cell line CHO) were stably transfected with the gene for APP-751 containing the double mutation Lys651 -Met652 to Asn651 -Leu652 (APP-751 numbering) commonly called the Swedish mutation using the method described in Citron et al., (1992) Nature 360:672-674. The transfected cell lines were designated as 293 751 SWE and CHO 751 SWE, and were plated in Corning 96 well plates at 2.5×104 or 1×104 cells per well respectively in Dulbecco's minimal essential media plus 10% fetal bovine serum. Following overnight incubation at 37°C in an incubator equilibrated with 10% carbon dioxide (CO2), the media were removed and replaced with 200 μL per well of media containing a test compound. After a two hour pretreatment period, the media were again removed and replaced with fresh media containing the test compound and the cells were incubated for an additional two hours.

Test compound stocks were prepared in DMSO such that at the final concentration used in the treatment, the concentration of DMSO did no exceed 0.5%. After treatment, plates were centrifuged in a Beckman GPR at 1200 rpm for five minutes at room temperature to pellet cellular debris from the conditioned media. From each well, 100 μL of conditioned media were transferred into an ELISA plate precoated with antibody 266 against βAP-13-28 (Seubert et al., supra.) and stored at 4°C overnight. An ELISA assay employing labeled antibody 6C6 (against βAP-1-16) was run the next day to measure the amount of Aβ produced.

Cytotoxic effects of the test compounds were measured by a modification of the method of Hansen et al., (1989) J. Immun. Meth. 119:203-210. To the cells remaining in the tissue culture plate, was added 25 μL of a 3, (4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide (MTT) stock solution (5 mg/mL) to a final concentration of 1 mg/mL. Cells were incubated at 37°C for one hour, and cellular activity was stopped by the addition of an equal volume of MTT lysis buffer (20% w/v sodium dodecylsulfate in 50% DMF, pH 4.7). Complete extraction was achieved by overnight shaking at room temperature. The difference in the OD562 nm and the OD650 nm was measured in a Molecular Devices UVmax microplate reader as an indicator of the cellular viability.

The results of the Aβ ELISA were fit to a standard curve and expressed as ng/mL Aβ peptide. In order to normalize for cytotoxicity, these Aβ results were divided by the MTT results and expressed as a percentage of the results from a drug -free control.

The test compounds were assayed for Aβ production inhibition activity in cells at 10 μg/mL using the test. The results presented in Table 1 are the mean and standard deviation of at least six replicate assays. IC50 means the concentration of test compound in micromoles/liter required to inhibit Aβ production by 50%.

TABLE 1
______________________________________
Aβ production inhibition activity in cells
##STR6##
Aβ Production Inhibition Activity is given as an IC50 (μM)
or
as a % inhibition at (@) 10 μg/ml or a given concentration of
compound in μg/ml.
______________________________________
In each of the following compounds, R1 represents 3-(4-t-
butylphenoxy)phenyl, Y1 represents N(OH), Z represent S and
Y2 represents NH.
No. L1 L2 R2 Inhibition
______________________________________
1 CH(CH3)
CH2 phenyl 11.2
2 CH(CH3)
CH2 3,4-dichlorophenyl
77%
3 CH(CH3)
CH2 4-chlorophenyl
11.7
4 CH(CH3)
CH2 4-fluorophenyl
11.1
5 CH(CH3)
CH2 2-methylphenyl
10.3
6 CH(CH3)
CH2 3-methylphenyl
11.1
7 CH(CH3)
CH2 4-methylphenyl
15.6
8 CH(CH3)
CH2 3-chloro-4- 9
methylphenyl
9 CH(CH3)
CH2 3,4-dimethylphenyl
62%
10 CH(CH3)
CH2 4-methoxyphenyl
>20
11 CH(CH3)
CH2 3,4-dimethoxyphenyl
31%
12 CH(CH3)
-- 1-naphthyl >20
13 CH(CH3)
CH(CH3)
phenyl 46%
14 CH(CH3)
CH(Ph) phenyl 44%
15 CH(CH3)
CH2 3,4- 28%
methylenedioxyphenyl
16 CH(CH3)
CH2 3-chlorophenyl
12
17 CH(CH3)
CH2 2,4-dichlorophenyl
5.7
18 CH(CH3)
CH2 3-fluorophenyl
18.3
19 CH(CH3)
n-hexyl 11
20 CH(C2 H5)
CH2 phenyl 3.6
21 CH(C2 H5)
CH2 3-methoxyphenyl
5.6
22 CH(C2 H5)
-- 2-chloro-3- 10%
trifluoromethylphenyl
23 CH(C2 H5)
-- 4-chloro-3- 11
trifluoromethylphenyl
24 CH(C2 H5)
n-hexyl 8.8
25 CH(C2 H5)
-- 4-(n-butoxy)phenyl
16
26 CH2 CH2 phenyl 3.4
27 CH2 n-hexyl 9.4
28 CH2 CH2 3,4-dichlorophenyl
4.2
29 CH(C3 H7)
CH2 phenyl 7.5
30 CH(C3 H7)
CH2 3-chlorophenyl
9.1
31 CH(C3 H7)
CH2 3,4-dichlorophenyl
10
32 CH(C3 H7)
CH2 3-methylphenyl
9.0
33 CH(benzyl)
CH2 phenyl 16% @
5 μg/ml
34 CH(benzyl)
n-hexyl 20% @
5 μg/ml
35 CH(n- -- 4-chloro-3- 9%
C6 H13) trifluoromethylphenyl
36 CH(n- CH2 phenyl 26%
C6 H13)
37 CH(n- n-hexyl 0%
C6 H13)
______________________________________
In each of the following compounds R1 represents 3-(4-t-
butylphenoxy)phenyl, Z represents S and Y2 represents NH.
No. L1 Y1 L2
R2 Inhibition
______________________________________
38 CH(CH3)
N(N(CH3)2)
CH2
phenyl 32%
39 CH(CH3)
NH CH2
phenyl 10.5
40 CH2 NH CH2
phenyl 9.1
41 CH2 NH CH2
4-chloro-3-
4.0
trifluoromethyl
phenyl
42 CH2 NH n-hexyl 17.2
43 CH2 N(OCH3)
n-hexyl 16%
44 CH2 N(CH3)
CH2
3,4- 40%
dichlorophenyl
______________________________________
In each of the following compounds L1 represents CH(CH3),
Y2
represents NH, L2 represents CH2 and R2 represents
phenyl.
No. R1 Y1 Z Inhibition
______________________________________
45 3-(4-n-butylphenoxy)phenyl
N(OH) S 9.2
46 3-(4-t-butylphenoxy)phenyl
N(OH) O 68%
47 3-phenoxyphenyl NH S 8%
______________________________________
In each of the following compounds L1 represents CH2, Y1
represents N(OH), Z represents S and Y2 represents NH.
No R1 L2 R2 Inhibition
______________________________________
48 3-(4-t-butylbenzyl-
CH2
3-methyl
29% @ 5
oxy)phenyl phenyl μg/ml
49 3-(4-t-butylbenzyl-
CH2
phenyl 6.7
oxy)phenyl CH2
phenyl
50 3-(4-t-butylbenzyl-
n-hexyl 25% @ 5
oxy)phenyl μg/ml
51 3-(4-t-butylbenzyl-
CH2
3,4- 38% @ 5
oxy)phenyl dichloro
μg/ml
phenyl
52 4-(4-t-butylbenzyl-
CH2
phenyl 22% @ 5
oxy)phenyl μg/ml
53 4-(4-2-phenyl-
CH2
phenyl 10% @ 5
benzyloxy)phenyl
CH2
phenyl μg/ml
54 4-(3,4-dichloro-
CH2
phenyl 25% @ 5
benzyloxy)phenyl μg/ml
55 4-(3,4-dichloro-
n-hexyl 37% @ 5
benzyloxy)phenyl μg/ml
56 4-(3,4-difluoro-
CH2
phenyl 28% @ 5
benzyloxy)phenyl μg/ml
57 4-(3,4-difluoro-
n-hexyl 4% @ 5
benzyloxy)phenyl μg/ml
58 4-(2,5-dimethyl-
CH2
phenyl 32% @ 5
benzyloxy)phenyl μg/ml
59 4-(2,5-dimethyl-
n-hexyl 28% @ 5
benzyloxymethyl μg/ml
60 4-(3,5-dimethoxy-
CH2
phenyl 17% @ 5
benzyloxy)phenyl μg/ml
61 4-(3,5-dimethoxy-
n-hexyl 28% @ 5
benzyloxy)phenyl μg/ml
62 4-(4-n-butylbenzyl-
CH2
phenyl 17% @ 5
oxy)phenyl μg/ml
63 4-(4-n-butylbenzyl-
n-hexyl 9% @ 5
oxy)phenyl μg/ml
64 4-(2,6-dichloro-
CH2
phenyl 18%
benzyloxy)phenyl
65 4-(2,6-dichloro-
CH2
3-methyl
7%
benzyloxy)phenyl phenyl
66 3-(4-butoxybenzyl-
CH2
phenyl 21%
oxy)phenyl
67 4-(4-acetyl-3-
CH2
phenyl 42%
hydroxy-2-propy-
benzyloxy)phenyl
68 4-(4-acetyl-3-
n-hexyl 19.7
hydroxy-2-propyl-
benzyloxy)phenyl
69 4-(4-benzoyl-3-
-- 4-chloro-
14%
hydroxy-2-propyl- 3-
benzyloxy)phenyl trifluoro-
methyl-
phenyl
70 3-(3-hydroxy-4-
n-hexyl 37%
propanoyl-2-propyl-
benzyloxy)phenyl
71 3-(3-hydroxy-4-
CH2
phenyl 12.0
propanol-2-propyl-
benzyloxy)phenyl
72 3-(3,5-di-t-butyl-
CH2
phenyl 8.1
4-methoxybenzyl-
oxy)phenyl
73 4-(3,5-di-t-butyl-
CH2
phenyl 5.9
4-methoxybenzyloxy)
phenyl
74 2-and-4-(2-phenyl-
CH2
phenyl 7.1
benzyloxy)phenyl (mixture)
______________________________________
In the following compounds L1 represents CH(CH3), Y1
represents N(OH), Z represents S and Y2 represents NH.
No. R1 L2 R2
Inhibition
______________________________________
75 3-(4-t- CH2
phenyl
8.9
butylbenzyloxy)phenyl
______________________________________
In the following compounds, L1 represents CH2, Y1
represents
N(OH), Z represents S and Y2 represents NH.
No. R1 L2 R2
Inhibition
______________________________________
76 4-biphenyl CH2
3,4- 13%
dichloro
phenyl
77 4-biphenyl CH2
phenyl 2% @ 5 μg/ml
78 4-biphenyl n-hexyl 24% @ 5 μg/ml
79 4-biphenyl CH2
3-methyl
15%
phenyl
80 4-(4-t- n-hexyl 11%
butylphenyl)
phenyl
81 4-(4-t- CH2
phenyl 20%
butylphenyl)
phenyl
82 4-(4-t- CH2
3-methyl
30%
butylphenyl) phenyl
phenyl
83 4-(4-t- -- 4- 12.4
butylphenyl) chloro-3-
phenyl trifluoro-
methyl-
phenyl
84 3-(4-t-butyl-
CH2
phenyl 15
benzamido)-
phenyl
85 3-(4-t-butyl-
n-hexyl 18.6
benzamido)-
phenyl
86 3-(4-t-butyl-
CH2
phenyl 5.9
phenylthio)
phenyl
87 3-(4-t-butyl-
n-hexyl 9.9
phenylthio)
phenyl
88 4-(4-t-butyl-
CH2
phenyl 7.5
phenylthio)
phenyl
89 4-(4-t-butyl-
n-hexyl 5.3
phenylthio)
phenyl
90 3-(4-t-butyl-
n-hexyl 22
phenyl
sulphonyl)
phenyl
91 3-(4-t-butyl-
CH2
phenyl 17.1
phenyl-
sulphinyl)
phenyl
92 3-(4-t-butyl-
CH2
phenyl 25%
phenyl
sulphonyl)
phenyl
93 3-(4-t-butyl-
n-hexyl 13%
phenyl
sulphonyl)
phenyl
94 4-(4-t-butyl-
CH2
phenyl 13.1
phenylthio
methyl)phenyl
95 4-(4-t-butyl-
-- 4- 4.6
phenylthio chloro-3-
methyl)phenyl trifluoro
methyl
phenyl
96 4-(4-1,1- n-hexyl 24.8
dimethylpropyl
phenoxymethyl)
phenyl
97 4-(4-1,1- CH2
phenyl 10.0
dimethylpropyl
phenoxymethyl)
phenyl
______________________________________
In the following compounds L1 represents CH(CH3), Y1
represents N(OH), Z represents S and Y2 represents NH.
No. R1 L2 R2
Inhibition
______________________________________
98 3-(4-t-butyl- n-hexyl 13
benzamido)phenyl
99 3-(4-t-butyl- CH2
phenyl 43% @ 5 μg)ml
benzamido)phenyl
100 3-(4-t-butyl- CH2
3-methyl
1% @ 5 μg)ml
benzamido)phenyl phenyl
______________________________________
In the following compounds, L1 represents CH2, Y1
represents
N(OH) and Y2 represents NH.
No. R1 Z L2
R2
Inhibition
______________________________________
101 3-(2-propyl-4-
S CH2
phenyl 4.5
t-butylphenoxy)
phenyl
102 3-(2-propyl-4-
S n-hexyl 6.1
t-butylphenoxy)
phenyl
103 3-(2-propyl-4-
O 4-chloro-3-CF3 -phenyl
20
t-butylphenoxy)
phenyl
______________________________________
In each of the following compounds, R1 represents 4-(4-t-
butylphenoxyphenyl), L1 represents a bond, Y1 represents NH,
Z represents S and Y2 represents NH.
No. L2 R2 Inhibition
______________________________________
104 CH2 2-chlorophenyl 13.4
105 CH2 3-chlorophenyl 9.4
106 CH2 4-chlorophenyl 9.2
107 CH2 3-bromophenyl 7.2
108 CH2 4-bromophenyl 8.9
109 CH2 2-methoxyphenyl 37%
110 CH2 3-methoxyphenyl 21.2
111 CH2 4-methoxyphenyl 8.30
112 CH2 3-fluorophenyl 7.60
113 CH2 4-fluorophenyl 5.60
114 -- phenyl 18.2
115 CH2 phenyl 16.4
116 (CH2)2
phenyl 19.8
117 (CH2)3
phenyl 17.0
118 (CH2)4
phenyl 16.4
119 -- cyclohexyl 15.2
120 CH2 2-pyridyl 15.6
121 CH2 3-pyridyl 37%
122 CH2 4-pyridyl 23.5
123 (CH2)2
2-pyridyl 12.3
124 CH2 n-t-butoxycarbonyl-2-
31%
piperidinyl
125 CH2 n-t-butoxycarbonyl-3-
9.30
piperidinyl
126 CH2 2-piperidinyl 7.00
127 CH2 3-piperidinyl 11.7
128 CH2 2,4-difluorophenyl
7.3
129 CH2 3,4-difluorophenyl
4.5
130 CH2 2-chloro-4-fluorophenyl
7.2
131 CH2 3-chloro-4-fluorophenyl
5.0
132 CH2 4-fluoro-2-trifluoromethyl-
8.0
phenyl
133 CH2 3,4-dichlorophenyl
6.5
134 CH2 2-trifluoromethyl-1,3,4-
thiadiazolyl 10.2
135 n-hexyl 10.6
______________________________________
In each of the following compounds, L1 represemts a bond, Y1
represents NH, Y2 represents NH and L2 represents a bond.
No. R2 R1 Z Inhibition
______________________________________
136 3,4-dichloro 5-quinolinyl O 17.8
phenyl
137 3,4-dichloro 2-benzothiazolyl
O 10.3
phenyl
138 3,4-dichloro-
2-pyrimidinyl O 7%
phenyl
139 3,4-dichloro 4-chlorophenyl S 6.6
phenyl
140 3,4-dichloro 2-benzimidazolyl
O 17.4
phenyl
141 3,4-dichloro 2-dimethylaminopropyl
O 7.5
phenyl thiophenyl
142 3,4-dichloro 2-methoxycarbonyl-
O 22%
phenyl phenyl
143 3,4-dichloro 2-(5,6-dimethyl)
O 14.9
phenyl benzimidazolyl
144 3,4-dichloro 2,4-dimethoxyphenyl
O 41%
phenyl
145 3,4-dichloro 4-(3,4-dichlorophenyl)
O 6.2
phenyl thiophenyl
146 3,4-dichloro benzyl S 21.8
phenyl
147 3,4-dichloro 3-trifluoromethyl
S 4.7
phenyl phenyl
148 3,4-dichloro 4-phenoxyphenyl
S 7.5
phenyl
149 3,4-dichloro 4-methylthiophenyl
O 13.8
phenyl
150 3,4-dichloro 3-bromo-4- O 5.2
phenyl methylthiophenyl
151 3,4-dichloro 2-chlorophenyl O 10.1
phenyl
152 3,4-dichloro phenyl O 11.4
phenyl
153 3,4-dichloro 3,4-dichlorophenyl
S 1.7
phenyl
154 3,4-dichloro 4-methylthiophenyl
O 8.6
phenyl
155 3,4,5- 4-methylthiophenyl
O 5.8
trichlorophenyl
156 4-chlorophenyl
4-methylthiophenyl
O 13%
157 2-chlorophenyl
4-methylthiophenyl
O 41%
158 2,3- 4-methylthiophenyl
O 51%
dichlorophenyl
159 2,4- 4-methylthiophenyl
O 4%
dichlorophenyl
160 4,5-dichloro-2-
4-methylthiophenyl
O 48%
methylphenyl
161 4-chlorophenyl
4-methylsulfonylphenyl
O 40%
162 3-chlorophenyl
3-bromo-4- O 5.7
methylthiophenyl
163 3-chlorophenyl
2,5-dichloro-4-
O 10.5
methylthiophenyl
164 4-methylthio 3-bromo-4- O 14.4
phenyl methylthiophenyl
165 4-chloro-3-CF3 -
3-methoxy-4- O 42%
phenyl methoxycarbonylphenyl
166 4-chloro-3-CF3 -
4-chloro-3- S 2.8
phenyl trifluoromethylphenyl
167 4-chloro-3-CF3 -
4-chloro-3- O 1.4
phenyl trifluoromethylphenyl
______________________________________
In each of the following compounds, Z represents S, Y2
represents NH, L2 represents a bond and R2 represents 4-
chloro-3-trifluoromethylphenyl.
No. R1 L1
Y1
Inhibition
______________________________________
167 4-(4-t-butylphenoxy)phenyl
-- NH 3.3
168 3-(4-t-butylphenoxy)phenyl
-- NH 3.9
169 4-(3-t-butylphenoxy)phenyl
-- NH 4.6
170 2-(4-t-butylphenoxy)phenyl
-- NH 4.4
171 2-(4-t-butylphenoxy)phenyl
-- N(OH) 10.8
172 3-(3-t-butylphenoxy)phenyl
-- NH 3.8
173 3-(3-t-butylphenoxy)phenyl
-- N(OH) 2.8
174 4-(2-t-butylphenoxy)phenyl
-- NH 66%
______________________________________
In each of the following compounds, L1 represents a bond, Y1
represents NH, Z represents S, Y2 represents NH and R2
represents 3-trifluoromethylphenyl.
No. R1 L2
Inhibition
______________________________________
175 4-(4-t-butylphenoxy)phenyl
-- 7.3
176 4-(3-t-butylphenoxy)phenyl
-- 4.6
177 4-(2-t-butylphenoxy)phenyl
-- 7.6
178 4-(3-t-butylphenoxy)phenyl
CH2
8.6
179 4-(4-t-butylphenoxy)phenyl
CH2
6.5
______________________________________
In each of the following compounds, R1 represents 2-(4-t-
butylphenoxyphenyl), Z represents S, and Y2 represents NH.
No. L1 Y1 L2
R2 Inhibition
______________________________________
180 CH2
N(OH) CH2
phenyl 11.6
181 -- NH CH2
phenyl 52%
182 CH2
N(OH) n-hexyl 34
183 -- NH CH2
3,4-dichlorophenyl
24%
184 -- NH CH2
3,4-dimethylphenyl
36%
185 -- NH n-hexyl 21%
______________________________________
In each of the following compounds Z represents S, Y1
represents NH, L2 represents CH2 and R2 represents
phenyl.
No. R1 L1 Y1
Inhibition
______________________________________
186 3-(4-t-butylphenoxy)phenyl
-- NH 9.0
187 3-(3-t-butylphenoxy)phenyl
CH2
N(OH) 12.1
188 4-(2-t-butylphenoxy)phenyl
-- NH 9.4
______________________________________
In each of the following compounds L1 represents a bond, Y1
represents NH, Z represents S, Y2 represents NH, L2
represents a bond a R2 represents 2-trilfuoromethyl-phenyl.
No. R1 Inhibition
______________________________________
189 4-(3-t-butylphenoxy)phenyl
37%
190 4-(2-t-butylphenoxy)phenyl
13.8 μM
______________________________________
In each of the following compounds L1 represents a bond, Y1
represents NH, Z represents S, Y2 NH, L2
represents a bond and R2 represents 4-chlorophenyl.
No. R1 Inhibition
______________________________________
191 4-(4-t-butylphenoxy)phenyl
33%
192 3-(4-t-butylphenoxy)phenyl
8.8
______________________________________
In each of the following compounds R1 represents 4-(4-t-
butyl-2-propyl)phenoxyphenyl, L1 represents a bond, Y1
represents NH, Z represents S and Y2 NH.
No. L2 R2 Inhibition
______________________________________
193 -- 4-chloro-3-trifluoromethylphenyl
29%
194 CH2 3-methylphenyl 31%
195 -- 3-trifluoromethylphenyl
11
196 -- 3,4-dimethylphenyl
44%
197 CH2 phenyl 4.6
______________________________________
In each of the following compounds, Z representsd S, Y2
represents NH, L2 represents CH2 and R2 represents
phenyl.
No. R1 L1 Y1
Inhibition
______________________________________
198 4-(4-t-butylphenoxy)phenyl
CH2
S 10
199 4-acetyl-3-hydroxy-2-
CH2
S 2
propylphenyl
200 3,4-dichlorophenyl
-- NH 21.8
______________________________________
In each of the following compounds R1 represents 4-benzoyl-3-
hydroxy-2-propylphenyl, L1 represents a bond, Y1 represents
NH, Z represents S and Y2 represents NH.
No. L2 R2 Inhibition
______________________________________
201 -- 3-trifluoromethylphenyl
20
202 n-hexyl 42%
203 -- 4-t-butylphenyl
2%
______________________________________
In each of the following compounds L1 represents a bond, Y1
represents NH, Y2 represents NH, L2 represents a bond and
R2
represents 4-chloro-3-trifluoromethylphenyl.
No. R1 Z Inhibition
______________________________________
204 4-(2-quinolylthio)phenyl
S 3.7
205 4-(4-ethoxycarbonyl-2-
S 3.1
quinolylthio)phenyl
206 4-(6-chloro-2-quinolylthio)phenyl
S 2.7
207 4-(4-phenyl-2-quinolylthio)phenyl
S >20
208 4-(6-methoxy-2-quinolylthio)phenyl
S 4.4
209 4-(4-quinolylthio)phenyl
S 4.7
210 4-(7-trifluoromethyl-4-
S 4.5
quinolylthio)phenyl
211 4-(2-naphthylthio)phenyl
S 2.6
212 4-(1-naphthylthio)phenyl
S 4.9
213 4-(2-benzimidazolylthio)phenyl
S 7.1
214 4-(2-benzoxazolylthio)phenyl
S 12.5
215 4-(2-benzothiazolylthio)phenyl
S 3.9
216 4-(5-chloro-2- S 1.7
benzothiazoylthio)phenyl
217 4-(2-quinolylsulfinyl)phenyl
S 12.1
218 4-(2-quinolylsulfonyl)phenyl
S 3.7
219 4-(2-quinolylamino)phenyl
S 11.8
220 4-(2-quinolyloxy)phenyl
S 9.7
221 4-(2-quinolylthio)phenyl
O >20
222 4-(6-phenyl-3-pyridazylthio)phenyl
S 4.4
223 4-(2-pyridazylthio)phenyl
S 8.3
224 4-(5-nitroquinolylthio)phenyl
S 4.3
225 4-(4-isopropylsulfonyl)phenyl
S 3.3
______________________________________
In each of the following compounds L1 represents a bond, Y1
represents NH, Y2 represents NH, L2 represents a bond and
R2
represents 3,4-dichlorophenyl.
No. R1 Z Inhibition
______________________________________
226 4-(2-quinolythio)phenyl
S 13.8
______________________________________
In each of the following compounds L1 represents a bond, Y1
represents a bond, Z represents O, Y2 represents NH, L2
represents a bond and R2 represents 3,4-dichlorophenyl.
No. R1 Inhibition
______________________________________
227 2-pyridyl 32%
228 6-1,2,5-triazinyl 22%
229 1-piperidinyl 8%
230 1-ethyl-3-piperidinyl 28%
231 2,2,6,6-tetramethyl-4-piperidinyl
40-51% @
2.5 μg/ml
232 5-chloro-1,3,4-thiadiazol-2-yl
2.0
233 2-methoxycarbonylbenzothiophen-3-yl
15.2
234 5-methyl-1,3,4-thiadiazol-2-yl
5.0
235 1,3,4-thiadiazol-2-yl 10.0
236 5-trifluoromethyl-1,3,4-thiadiazol-2-yl
4.2
237 5-t-butyl-1,3,4-thiadiazol-2-yl
2.9
238 5-(2-chloro-1,1-dimethylethyl)-1,3,4-
1.4
thiadiazol-2-yl
239 5-hexylthio-1,3,4-thiadiazol-2-yl
1.4
240 5-bromo-1,3,4-thiadiazol-2-yl
1.8
241 2-oxazolyl -8%
242 5-methylisoxazol-3-yl 29%
243 2-thiazolyl 19.4
244 4,5-dihydrothiazolyl 10%
245 3-t-butylisoxazol-5-yl
9.8
246 3-t-butyl-1,2,4-oxadiazol-5-yl
5.8
247 3-trifluoromethyl-1,2,4-oxadiazol-5-yl
4.4
______________________________________
In each of the following compounds, R1 represents 5-methyl-
1,3,4-thiadiazol-2-yl, L1 represents a bond, Y1 represents
NH, Z represents O, Y2 represents NH and L2 represents a
bond.
No. R2 Inhibition
______________________________________
248 3-chlorophenyl 47%
249 2-fluorophenyl 6%
250 3-chloro-4-fluorophenyl
9.8
251 2,6-dimethylphenyl
-13%
252 3-methylthiophenyl
25%
______________________________________
In each of the following compounds R1 represents 5-bromo-
1,3,4-thiadiazol-2-yl, L1 represents a bond, Z represents O,
Y2 represents NH and L2 represents a bond.
No. Y1 R2 Inhibition
______________________________________
253 NH methyl 2%
254 N(CH3)
methyl 11%
______________________________________
In each of the following compounds, R1 represents 1,3,4-
thiadiazol-2-yl, L1 represents a bond, Y1 represents NH, Z
represents O, Y2 represents NH and L2 represents a bond.
No. R2 Inhibition
______________________________________
255 phenyl 1%
256 4-chlorophenyl
42%
______________________________________

The compounds of the present invention can be administered for prophylactic and/or therapeutic treatment of diseases related to the deposition of Aβ, such as Alzheimer's disease, Down's syndrome, and advanced aging of the brain. In therapeutic applications, the compounds are administered to a host already suffering from the disease. The compounds will be administered in an amount sufficient to inhibit further deposition of senile plaques. The specific dose of compound administered according to this invention to obtain therapeutic and/or prophylactic effects will, of course, be determined by the particular circumstances surrounding the case, including, for example, the compound administered, the route of administration, the condition being treated, the individual being treated and the like. A typical daily dose will contain a dosage level of from about 0.01 mg/kg to about 50 mg/kg of body weight of an active compound of this invention. Preferred daily doses generally will be from about 0.05 mg/kg to about 20 mg/kg, for example from about 0.1 mg/kg to about 10 mg/kg.

For prophylactic applications, the compounds of the present invention are administered to a warm-blooded mammal susceptible to Alzheimer's Disease or a βAP-related disease, but not already suffering from such disease. Such hosts may be identified by genetic screening and clinical analysis, as described in the medical literature. See e.g., Goate, (1991) Nature 349:704-706. The compounds will be able to inhibit or prevent the formation of senile plaques at a symptomatically early stage, preferably preventing even the initial stages of the μamyloid disease.

The compounds can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular and intranasal. The compounds of the present invention are preferably formulated prior to administration.

The active ingredient in such formulations comprises from 0.1% to 99.9% by weight of the formulation. By "pharmaceutically acceptable" it is meant that the carrier, diluent or excipient is compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.

For example, a typical pharmaceutical composition for intramuscular injection would contain about one μg to one mg of the compound in from one to four milliliters of sterile buffered water. The typical pharmaceutical composition for intravenous infusion would contain about one to one hundred milligrams of the compound in from one hundred to five hundred milliliters of sterile Ringer's solution.

The pharmaceutical formulations are prepared by known procedures using known and readily available ingredients. In making the compositions of the present invention, the active ingredient will usually be admixed with a carrier, or diluted by a carrier, or enclosed within a carrier which may be in the form of a capsule, sachet, paper or other container. When the carrier serves as a diluent, it may be solid, semi-solid or liquid material which acts as a vehicle, excipient or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols, (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, sterile packaged powders and the like.

Some examples of suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum, acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water syrup, methyl cellulose, methyl and propyl hydroxybenzoates, talc, magnesium stearate, and mineral oil. The formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents, or flavoring agents. Compositions of the invention may be formulated so as to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art.

The compositions are preferably formulated in a unit dosage form, each dosage containing from about 5 mg to about 500 mg, more preferably about 25 mg to about 300 mg of the active ingredient. The term "unit dosage form" refers to a physically discrete unit suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical carrier, diluent, or excipient. The following formulation examples are illustrative only and are not intended to limit the scope of the invention in any way.

______________________________________
Formulation 1
Hard gelatin capsules are prepared using the following
ingredients:
Quantity
(mg/capsule)
______________________________________
Active Ingredient 250
Starch, dried 200
Magnesium stearate 10
Total 460 mg
______________________________________

The above ingredients are mixed and filled into hard gelatin capsules in 460 mg quantities.

______________________________________
Formulation 2
A tablet is prepared using the ingredients below:
Quantity
(mg/tablet)
______________________________________
Active Ingredient 250
Cellulose, microcrystalline
400
Silicon dioxide, fumed 10
Stearic acid 5
Total 665 mg
______________________________________

The components are blended and compressed to form tablets each weighing 665 mg.

______________________________________
Formulation 3
An aerosol solution is prepared containing the following
components:
Weight %
______________________________________
Active Ingredient 0.25
Ethanol 29.75
Propellant 22 70.00
(chlorodifluoromethane)
Total 100.00
______________________________________

The active compound is mixed with ethanol and the mixture added to a portion of the Propellant 22, cooled to -30°C and transferred to a filling device. The required amount is then fed to a stainless steel container and diluted with the remainder of the propellant. The valve units are then fitted to the container.

______________________________________
Formuation 4
Tablets each containing 60 mg of active ingredient are made
as follows:
______________________________________
Active Ingredient 60 mg
Starch 45 mg
Microcrystalline cellulose
35 mg
Polyvinylpyrrolidone 4 mg
Sodium carboxymethyl starch
4.5 mg
Magnesium stearate 0.5 mg
Talc 1 mg
Total 150 mg
______________________________________

The active ingredient, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed with the resultant powders which are then passed through a No. 14 mesh U.S. sieve. The granules so produced are dried at 50°C and passed through a No. 18 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and talc, previously passed through a No. 60 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.

______________________________________
Formulation 5
Capsules each containing 80 mg medicament are made as
follows:
______________________________________
Active Ingredient 80 mg
Starch 59 mg
Microcrystalline cellulose
59 mg
Magnesium stearate 2 mg
Total 200 mg
______________________________________

The active ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 45 sieve, and filled into hard gelatin capsules in 200 mg quantities.

______________________________________
Formulation 6
Suppositories each containing 225 mg of active ingredient may
be made as follows:
______________________________________
Active Ingredient 225 mg
2,000 mg
Total 2,225 mg
______________________________________

The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2 g capacity and allowed to cool.

______________________________________
Formulation 7
Suspensions each containing 50 mg of medicament per 5 ml dose
are made as follows:
______________________________________
Active Ingredient 50 mg
Sodium carboxymethyl cellulose
50 mg
Syrup 1.25 ml
Benzoic acid solution 0.10 ml
Flavor q.v.
Color q.v.
Purified water to total 5 ml
______________________________________

The medicament is passed through a No. 45 mesh U.S. sieve and mixed with the sodium carboxymethyl cellulose and syrup to form a smooth paste. The benzoic acid solution, flavor and color are diluted with some of the water and added, with stirring. Sufficient water is then added to produce the required volume.

______________________________________
Formulation 8
An intravenous formulation may be prepared as follows:
______________________________________
Active Ingredient 100 mg
Mannitol 100 mg
5 N Sodium hydroxide 200 ml
Purified water to total
5 ml
______________________________________

Frequently, it will be desirable or necessary to introduce the pharmaceutical compositions directly or indirectly to the brain. Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier. Indirect techniques, which are generally preferred, involve formulating the compositions to provide for drug latentiation by the conversion of hydrophilic drugs into lipid-soluble drugs. Latentiation is generally achieved through blocking of the hydroxyl, carboxyl, and primary amine groups present on the drug to render the drug more lipid-soluble and amenable to transportation across the blood-brain barrier. Alternatively, the delivery of hydrophilic drugs can be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the blood-brain barrier.

The following examples illustrate the invention:

In this specification, the following abbreviations have been used. THF refer to tetrahydrofuran, DMF refers to dimethylformamide, HPLC refers to high pressure liquid chromatography, DMAP refers to 4-dimethylaminopyridine, and DBU refers to 1,8-diazabicyclo[5,4,0]undec-7-ene.

General Procedures

In each of the following examples, one of the following three general procedures was used.

Procedure A. An amine (1 equivalent), isothiocyanate (1 equivalent), and 4-dimethylaminopyridine (1.1 equivalent) were dissolved in THF (2.5 ml/mmole) and stirred for 16 hr at room temperature under an atmosphere of nitrogen. The solution was concentrated in vacuo, ethyl acetate added, and washed twice with water. The organic layer was dried over sodium sulfate and condensed. The thiourea was purified by HPLC over silica gel eluted with 30% ethyl acetate/hexane.

Procedure B. An amine (1 equivalent), isothiocyanate (1 equivalent), and potassium carbonate (1 equivalent) were dissolved in THF (2.5 ml/mmole) and refluxed for 3 hr. The cooled solution was concentrated, ethyl acetate added, washed with water, dried over sodium sulfate and condensed. The product was purified by HPLC over silica gel eluted with ethyl acetate/hexane.

Procedure C. An amine (1 equivalent) and isothiocyanate (1 equivalent) were dissolved in THF (2.5 ml/mmole) and stirred at room temperature for 16 hr. The solution was concentrated, ethyl acetate added, washed with water, dried over sodium sulfate, and concentrated. The product was purified by HPLC over silica gel eluted with ethyl acetate/hexane.

PAC 1-[4-(2-quinolyl)thiophenyl]-3-(4-chloro-3-trifluoromethylphenyl)thiourea

2-(4-aminophenylthio)quinoline(3.9 mmoles, 1.0 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (3.9 mmoles, 0.92 g) were reacted according to procedure A to yield 1.06 g, 56% of the title compound. Mass spec (FD) 490. Calculated for C23 H15 ClF3 N3 S2 : C, 56.38; H, 3.09, N, 8.58. Found: C, 56.60; H, 3.11; N, 8.42.

The necessary amine starting material was prepared as follows:

2-Chloroquinoline (0.04 moles, 6.54 g), 4-aminothiophenol (0.04 moles, 5.0 g) and potassium carbonate (0.04 moles, 5.52 g) were stirred at room temperature in 200 ml of ethanol for 18 hr. The reaction mixture was concentrated, ethyl acetate was added and the solution washed with water, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with ethyl acetate/hexane to yield 2-(4-aminophenylthio)quinoline 4.0 g, 40%. Mass Spec (FD) 252. Calculated for C15 H12 N2 S: C, 71.40; H, 4.79, N, 11.10. Found: C, 71.11; H, 4.98N, 11.20.

PAC 1-[4-(4-ethoxycarbonyl-2-quinolylthio)phenyl]-3-(4-chloro-3-trifluoromethyl phenyl)thiourea

2-(4-aminophenylthio)-4-ethoxycarbonylquinoline (6.0 mmoles, 2.0 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (6.0 mmoles, 1.4 g) were reacted according to procedure C to yield 3.0 g, 89% of the title compound. Mass Spec (FD) 561. Calculated for C26 H19 ClF3 N3 O2 S2 : C, 55.56; H, 3.41; N, 7.48. Found: C, 55.38H, 3.38; N, 7.36.

The necessary amine stating material was prepared as follows:

2-Chloro-4-ethoxycarbonylquinoline (0.024 moles, 5.56 g), 4-aminothiophenol (0.024 moles, 3.0 g) and 4-dimethylaminopyridine (0.024 moles, 2.9 g) were stirred in 250 ml ethanol for 3 days.

The reaction mixture was filtered, concentrated, ethyl acetate added, washed with water and dried over sodium sulfate. The solution was concentrated and the product purified by HPLC over silica gel eluted with 25% ethyl acetate/hexane to yield 2-(4-aminophenylthio)-4-ethoxycarbonylquinoline 2.8 g, 36%. (FD) 324. Calculated for C18 H16 N2 O2 S: C, 66.64; H, 4.97; N, 8.63. Found: C, 66.50; H, 5.00N, 8.54.

PAC 1-[4-(6-Chloro-2-quinolylthio)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)t hiourea

2-(4-Aminophenylthio)-6-chloroquinoline (2.9 mmoles, 0.83 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (3.2 mmoles, 0.76 g) were reacted according to procedure C to yield 0.78 g, 51% of the title compound. Mass Spec (FD) 524. Calculated for C23 H14 ClF3 N3 S2 : C, 52.68; H, 2.69; N, 8.01. Found: C, 52.66; H, 2.78; N, 7.91. M Pt 138°-1390°C

The necessary amine starting material was prepared as follows:

2,6-Dichloroquinoline (15.2 mmoles, 3.0 g), 4-aminothiophenol (15.2 mmoles, 1.9 g), and DMAP 15.2 mmoles, 1.85 g) were stirred at room temperature for 3 days. The solvent was removed, diluted with ethyl acetate, washed with water, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with 25% ethyl acetate in hexane to yield 2-(4-aminophenylthio)-6-chloroquinoline 830 mg, 19%. Mass spec (FD) 286. Calculated. for C15 H11 ClN2 S: C, 62.82H, 3.87; N, 9.77. Found: C, 63.09; H, 3.94; N, 9.61.

PAC 1-[4-(4-Phenyl-2-quinolylthio)phenyl]-3-(4-chloro-3-trifluoromethyl-phenyl) thiourea

2-(4-aminophenylthio)-4-phenylquinoline (3.0 mmoles, 1.0 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (3.3 mmoles, 0.79 g) were reacted according to procedure C to yield the title compound 1.03 g, 61%. Mass Spec (FD) 565. Calculated for C29 H19 ClF3 N3 S2 : C, 61.53; H, 3.38; N, 7.42. Found: C, 61.82; H, 3.63; N, 7.43.

The necessary amine starting material was prepared as follows:

2-Chloro-4-phenylquinoline (4.18 mmoles, 10.0 g), 4-aminothiophenol (41.8 mmoles, 5.2 g) and DMAP (41.8 mmoles, 5.0 g) were stirred in 200 ml ethanol and 50 ml THF for 3 days. The solution was concentrated, ethyl acetate added, washed with water, dried over sodium sulfate and the solvent removed. The product was purified by HPLC over silica gel eluted with 30% ethyl acetate in hexane to yield 2-(4-aminophenylthio)-4-phenylquinoline 6.2 g, 45%. Mass Spec (FD) 328. Calculated for C21 H16 N2 S: C, 76.80; H, 4.91N, 8.53. Found: C, 77.04; H, 5.00; N, 8.55.

PAC 1-[4-(6-Methoxy-2-quinolylthio)phenyl]-3-(4-chloro-3-trifluoromethylphenyl) thiourea

2-(4-aminophenylthio)-6-methoxyquinoline 15.0 mmoles, 4.2 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate 15.0 mmoles, 3.6 g) were reacted according to procedure C to yield 5.6 g, 72% of the title compound. Mass Spec (FD) 519. Calculated for C24 H17 ClF3 N3 OS2 : C, 55.44; H, 3.30; N, 8.08. Found: C, 55.62; H, 3.43; N, 8.27.

The necessary amine starting material was prepared as follows:

2-Chloro-6-methoxyquinoline (52.0 mmoles, 10.0 g), 4-aminothiophenol (52.0 mmoles, 6.5 g) and dimethylaminopyridine (52.0 mmoles, 6.3 g) were stirred for 16 hr in 250 ml ethanol. The reaction was condensed and purified by HPLC over silica gel eluted with 25-30% ethyl acetate/hexane to yield 2-(4-aminophenylthio)-6-methoxyquinoline. 8.5 g, 58% product. Mass spec (FD) 282. Calculated for C16 H14 N2 OS: C, 68.06; H, 5.00, N, 9.92. Found: C, 68.04; H, 4.97; N, 10.02.

PAC 1-[4-(4-Quinolylthio)Phenyl]-3-(4-chloro-3-trifluoromethylphenyl)thiourea

4-(4-aminophenylthio)quinoline (3.9 mmoles, 1.0 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (3.9 mmoles, 0.92 g) were reacted according to procedure A to yield of the title compound 1.06 g, 56%. Mass spec (FD) 490. Calculated for C23 H15 ClF3 N3 S2 : C, 56.38; H, 3.09, N, 8.58. Found: C, 56.60; H, 3.11; N, 8.42.

The necessary amine starting material was prepared as follows:

a) 4-Chloroquinoline (32.3 mmoles, 5.2 g), 4-nitrothiophenol (32.3 mmoles, 5.0 g) and potassium carbonate (32.3 mmoles, 4.46 g) were stirred at room temperature in 500 ml of ethanol for 16 hr. The reaction mixture was then concentrated, ethyl acetate was added and the solution washed with water, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with 20-40% ethyl acetate/hexane to yield 4-(4-nitrophenylthio)quinoline 4.9 g, 54%. Mass Spec (FD) 282. Calculated for C15 H10 N2 O2 S: C, 63.82H, 3.57N, 9.92. Found: C, 65.09; H, 3.81; N, 10.02.

b) 4-(4-nitrophenylthio)quinoline 17.0 mmoles, 4.9 g) was dissolved in 200 ml ethanol and hydrogenated over 5 g of 5% Pd/C at 40 psi for 1 hr at room temperature. The solution was filtered through celite and the solvent removed to yield 4-(4-aminophenylthio)quinoline 2.5 g, 58%. Mass spec (FD) 253. Calculated for C15 H12 N2 S: C, 71.40; H, 4.79, N, 11.10. Found: C, 71.12; H, 4.93N, 10.88.

PAC 1-[4-(7-Trifluoromethyl-4-quinolylthio)phenyl]-3-(4-chloro-3-trifluoromethy lphenyl)thiourea

4-(4-aminophenylthio)-7-trifluoromethylquinoline (0.01 mole, 3.2 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (0.01 mole, 2.4 g) were reacted according to procedure C to yield the title compound 3.5 g, 63%. Mass Spec (FD) 557. Calculated for C24 H14 ClF6 N3 S2 : C, 51.66; H, 2.53; N, 7.53. Found: C, 51.92; H, 2.53; N, 7.46.

The necessary amine starting material was prepared as follows:

4-Chloro-7-trifluoromethylquinoline (0.043 moles, 10.0 g), 4-aminothiophenol (0.043 moles, 5.3 g) and 4-dimethylaminopyridine (0.04 moles, 4.9 g) were stirred in 250 ml ethanol for 3 days. The reaction mixture was filtered, concentrated, ethyl acetate added, washed with water and dried over sodium sulfate. The solution was concentrated and the product purified by HPLC over silica gel eluted with 25% ethyl acetate/hexane to yield 4-(4-aminophenylthio)-7-trifluoromethylquinoline 10.2 g, 74%. Mass Spec (FD) 320. Calculated for C16 H11 F3 N2 S: C, 59.99; H, 3.46; N. 8.74. Found: C, 60.08; H, 3.49; N, 8.77.

PAC 1-[4-(2-Naphthylthio)phenyl]-3-(4-chloro -3-trifluoromethylphenyl)thiourea

2-(4-Aminophenylthio)naphthalene (4.2 mmoles, 1.06 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (4.2 mmoles, 0.93 g) were reacted according to procedure A to yield of the title compound 0.7 g, 34%. Mass Spec (FD) 489. Calculated for C24 H16 ClF3 N2 S2 : C, ; H, N, . Found: C, ; H, ; N, .

The necessary amine starting material was prepared as follows:

a) 2-Bromonaphthalene (0.08 moles, 16.6 g), 4-nitrothiophenol (0.08 moles, 12,4 g), potassium carbonate (0.1 moles, 13.4 g), copper bronze (6.0 g) and cuprous chloride (0.02 moles, 2.0 g) were heated under reflux for 4 days in 300 ml pyridine. The solution was filtered hot, concentrated, dissolved in ethyl acetate and washed thoroughly with 2N HCl and water. The solution was dried over sodium sulfate, concentrated and purified by HPLC over silica gel eluted with 5% ethyl acetate in hexane. The product was recrystallized from ethyl acetate and hexane to yield 2-(4-nitrophenylthio)naphthalene 2.6 g, 12% in the first crop. Mass spec. (FD) 281.

b) 2-(4-nitrophenylthio)naphthalene (9.2 mmoles, 2.6 g) was dissolved in 50 ml ethanol and 20 ml ethyl acetate and hydrogenated for 16 hrs at room temperature over 1.0 g 5% Pd/C. The solution was filtered through celite and concentrated to yield 2-(4-aminophenylthio)naphthalene 1.1 g, 47%.

PAC 1-[4-(1-Naphthylthiophenyl)-3-(4-chloro-3-trifluoromethylphenyl)thiourea

1-(4-aminophenylthio)naphthalene (20 mmoles, 5.0 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (20 mmoles, 4.7 g) were reacted according to procedure C to yield of the title compound, 2.0 g, 20%. Mass spec (FD) 488. Calculated for C24 H16 ClF3 N2 S2 : C, 58.95; H, 3.30, N, 5.73 Found: C, 59.17; H, 3.51; N, 5.91.

The necessary amine starting material was prepared as follows:

a) 1-Bromonaphthalene (0.15 mole, 31.0 g), 4-nitrothiophenol(0.11 mole, 17.0 g), potassium carbonate (0.15 moles, 20.7 g), copper bronze (0.3 moles, 18.9 g) and cuprous chloride (0.06 moles, 6.0 g) were refluxed in 500 ml pyridine for 3 days. The reaction was filtered while hot, and then concentrated. Ethyl acetate was added and the solution washed with water and 5N HCl, dried over sodium sulfate and concentrated. The product was purified twice by HPLC over silica gel eluted with 20% ethyl acetate/hexane followed by 5% ethyl acetate/hexane to yield 1-(4-nitrophenylthio)naphthalene 11.0 g, 26%. Mass Spec (FD) 282. Calculated for C16 H11 NO2 S: C, 68.31H, 3.94, N, 4.98. Found: C, 68.52; H, 4.00; N, 4.77.

b) 1-(4-Nitrophenylthio)naphthalene (39.0 mmoles, 11.0 g) was dissolved in 200 ml ethyl acetate and hydrogenated over 8 g of 5% Pd/C at 40 psi for 1 hr at room temperature. The solution was filtered through celite and the solvent removed to yield 1-(4-aminophenylthio)naphthalene 9.0 g, 92%. Mass spec (FD) 251. Calculated for C15 H17 NS: C, 76.46; H, 5.21, N, 5.57 Found: C, 76.71; H, 5.39; N, 5.47.

PAC 1-[4-(2-Benzimidazolylthio)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)thio urea

2-(4-Nitrophenylthio)benzimidazole (5.0 mmoles, 1.37 g) was reduced in ethyl acetate over 1.37 g 5% Pd/C, filtered and condensed and then reacted with 4-chloro-3-trifluoromethylphenylisothiocyanate (5.0 mmoles, 1.0 g) according to procedure C to yield the title compound 0.12 g, 5% product. Mass Spec (FD) 479. Calculated for C21 H14 ClF3 N4 S2 : C, 52.66; H, 2.95; N, 11.70. Found: C, 52.85; H, 3.18; N, 10.47.

The necessary starting material was prepared as follows:

2-Chlorobenzimidazole (0.033 moles, 5.0 g), 4-nitrothiophenol (0.033 moles, 4.14 g), and DBU (0.033 moles, 5.0 g) in 200 ml ethanol were heated at 55°C for 3 hr. The reaction was concentrated, ethyl acetate added, washed with water, dried over sodium sulfate and the solvent removed. The solid was slurred in hexane to yield 2-(4-nitrophenylthio)benzimidazole 1.37 g, 15%. Mass Spec (FD) 271. Calculated for C13 H8 N3 O2 S: C, 57.55; H, 3.34; N, 15.49. Found: C, 57.50; H, 5.54; N, 15.23.

PAC 1-[4-(2-Benzoxazolylthio)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)thiour ea

2-(4-Aminophenylthio)benzoxazole (10.0 mmoles, 2.6 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (10.0 mmoles, 2.4 g) were reacted according to procedure A to yield of the title compound 1.6 g, 33%. Mass Spec (FD) 479. Calculated for C21 H13 ClF3 N3 OS2 : C, 52.56; H, 2.73; N, 8.75. Found: C, 52.47; H, 2.70; N, 8.48.

The necessary amine starting material was prepared as follows:

2-Chlorobenzoxazole (0.065 moles, 10.0 g), 4-aminothiophenol (0.065 moles, 8.1 g) and potassium carbonate (0.065 moles, 9.0 g) were stirred for 3 days at room temperature in 250 ml ethanol, filtered, and concentrated. Ethyl acetate was added and the solution washed with water, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with 50% ethyl acetate in hexane to yield 2-(4-aminophenylthio)benzoxazole 12.5 g, 79%. Mass spec (FD) 243. Calculated for C11 H10 N2 OS: C, 64.44; H, 4.16; N, 11.56. Found: C, 64.53; H, 4.22; N, 11.58.

PAC 1-[4-(2-Benzothiazolylthio)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)thio urea

2-(4-Aminophenylthio)benzothiazole (10.0 mmoles, 2.6 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate 10.0 mmoles, 2.4 g) were reacted according to procedure C to yield of the title compound, 1.5 g, 30%. Mass Spec (FD) 495. Calculated for C21 H13 ClF3 N3 S3 : C, 50.85H, 2.64N, 8.47. Found: C, 51.02; H, 2.71; N, 8.31.

The necessary amine starting material was prepared as follows:

2-Chlorobenzothiazole (0.06 moles, 10.1 g), 4-aminothiophenol (0.06 moles, 7.5 g) and potassium carbonate (0.06 moles, 8.3 g) were stirred for 3 days at room temperature in 250 ml ethanol, filtered, and concentrated. Ethyl acetate was added and the solution washed with water, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with 40-60% ethyl acetate in hexane to yield 2-(4-aminophenylthio)benzothiazole 13.7 g, 88%. Mass spec (FD) 258. Calculated for C11 H10 N2 S2 : C, 60.44; H, 3.90; N, 10.84. Found: C, 60.63; H, 3.98; N, 11.01.

PAC 1-[4-(5-Chloro-2-benzothiazolylthio)phenyl]-3-(4-chloro-3-trifluoromethylph enyl)thiourea

2-(4-aminophenylthio)-5-chlorobenzothiazole (5.5 mmoles, 1.6 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (5.5 mmoles, 1.3 g) were reacted according to procedure C to yield the title compound 0.7 g, 24%. Mass Spec (FD) 529. Calculated for C21 H12 Cl2 F3 S3 : C, 47.55; H, 2.28; N, 7.92. Found: C, 47.55; H, 3.32; N, 7.87.

The necessary amine starting material was prepared as follows:

2,5-Dichlorobenzothiazole (0.049 moles, 10.0 g), 4-aminothiophenol (0.049 moles, 6.1 g) and DMAP (0.049 moles, 6.0 g) were stirred for 3 days at room temperature in 250 ml ethanol, filtered, and concentrated. Ethyl acetate was added and the solution washed with water, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with 25% ethyl acetate in hexane to yield 2-(4-aminophenylthio)-5-chlorobenzothiazole 3.2 g, 22%. Mass spec (FD) 292. Calculated for C13 H9 ClN2 S2 : C, 53.33; H, 3.10; N, 9.57. Found: C, 53.03; H, 3.30; N, 9.70.

PAC 1-[4-(2-Quinolylsulfinyl)phenyl]-3-(4-chloro -3-trifluoromethylphenyl)thiourea

2-(4-aminophenylsulfinyl)quinoline (4.8 mmoles, 1.3 g) and 4-chloro-3-trifluoromethylphenylisocyanate (4.8 mmoles, 1.15 g) were reacted according to procedure C to yield the title compound 0.4 g, 16%. Mass Spec (FD) 505. Calculated for C23 H15 ClF3 OS2 : C, 58.29; H, 3.19; N, 8.87. Found: C, 58.59; H, 3.60; N, 8.27.

The necessary starting material was prepared as follows:

a) 2-(4-Nitrophenylthio)quinoline (8.8 mmoles, 2.48 g) and m-chloroperbenzoic acid (8.8 mmoles, 3.37 g) were dissolved in 100 ml methylene chloride and stirred at room temperature for 18 hr. The solution was washed with water, dried over sodium sulfate, and concentrated. The product was purified by HPLC over silica gel eluted with 40% ethyl acetate/hexane to yield 4-nitrophenyl-2-quinolylsulfone 1.5 g, 54% and 4-nitrophenyl-2-quinolylsulfoxide 0.82 g, 31%. Sulfone: Mass spec (FD) 314. Calculated for C15 H10 N2 O4 S: C, 57.32; H, 3.21; N, 8.91. Found: C, 57.37; H, 3.18; N, 8.72. Sulfoxide: Mass spec (FD) 266. Calculated for C15 H10 N2 O3 S: C, 60.39; H, 3.38; N, 9.39. Found: C, 60.64; H, 3.18; N, 8.72.

b) 4-Nitrophenyl-2-quinolylsulfone (9.0 mmoles, 2.7 g) was dissolved in 200 ml ethanol and 75 ml DMF and was then hydrogenated over 2.2 g 5% Pd/C at room temperature for 3 hr. The reaction mixture was then filtered through celite and condensed to afford 2-(4-aminophenylsulfinyl)quinoline

PAC 1-[4-(2-Quinolylsulfonyl)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)thiour ea

2-(4-Aminophenylsulfonyl)quinoline (5.3 mmoles) and 4-chloro-3-trifluoromethylphenylisocyanate (5.8 mmoles, 1.38 g) were reacted according to procedure A to yield the title compound, 0.6 g, 2%. Mass Spec (FD) 521. Calculated for C23 H15 ClF3 N3 O2 S2 : C, 52.93; H, 2.90; N, 8.05. Found: C, 53.21; H, 3.09; N, 8.25.

The necessary amine starting material was prepared as follows:

4-Nitrophenyl-2-quinolylsulfoxide (5.3 mmoles, 1.5 g) prepared as described in Example (14, step a), was dissolved in ethanol/ethyl acetate and hydrogenated over 1 g 5% Pd/C at room temperature for 3 hr. The reaction mixture was then filtered through celite and condensed to afford 2-(4-aminophenylsulfonyl)quinoline Mass spec (FD) 284. Calculated for C15 H12 N2 O2 S: C, 63.36; H, 4.25; N, 9.85. Found: C, 62.16; H, 4.25; N, 9.25.

PAC 1-[4-(2-quinolylamino)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)thiourea

2-(4-aminophenylamino)quinoline (5.0 mmoles, 1.15 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (5.0 mmoles, 1.20 g) were reacted according to procedure C of to yield the title compound 1.60 g, 68%. Mass Spec (FD) 473. Calculated for C23 H16 ClF3 N4 S: C, ; H, ; N, . Found: C, ; H, ; N, .

PAC 1-[4-(2-Quinolyloxy)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)thiourea

2-(4-Aminophenoxy)quinoline (2.4 mmoles, 0.56 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (2.4 mmoles, 0.57 g) were reacted according to procedure C to yield the title compound, 0.8 g. 70%. Mass Spec (FD) 473. Calculated for C23 H15 ClF3 N3 OS: C, 58.29; H, 3.19; N, 8.87. Found: C, 58.47; H, 3.48; N, 8.85.

The necessary amine starting material was prepared as follows:

a) 4-Nitrophenol(41 mmoles, 5.1 g) was dissolved in DMF (500 ml) and treated with sodium hydride (41 mmoles previously washed with hexane). After stirring at room temperature for 1 hr, 2-chloroquinoline (40 mmoles, 6.5 g) was added dropwise and stirred for 16 hr at room temperature followed by refluxing for 6 hr. Water was added to the cooled solution and the product was extracted with ethyl acetate. The ethyl acetate solution was washed with water, and 5N NaOH, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with ethyl acetate/hexane to yield 2-(4-nitrophenoxy)quinoline. 3.1 mmoles, 8%. Mass Spec (FD) 266. Calcd for C15 H10 N2 O3 : C 67.45; H, 3.84; N, 10.40. Found: C, 65.84; H, 3.88; N, 8.47.

b) 2-(4-Nitrophenoxy)quinoline(3 mmoles, 0.82 g) was dissolved in 100 ml ethyl acetate and hydrogenated over 2.0 g 5% Pd/C at room temperature for 1 hr. The solution was filtered through cellulose and concentrated to yield 2-(4-aminophenoxy)quinoline 0.58, 82%. Mass Spec (FD) 236. Calculated for C15 H12 N2 O: C, 76.25; H, 5.12, N, 11.86. Found: C, 75.97; H, 5.37; N, 11.83.

PAC 1-[4-(2-Quinolylthio)phenyl]-3-(4-chloro-3-trifluoromethylphenyl)urea

2-(4-Aminophenylthio)quinoline (3.9 mmoles, 1.0 g) and 4-chloro-3-trifluoromethylphenylisocyanate (3.9 mmoles, 0.85 g) were reacted according to procedure A to yield the title compound, 0.8 g, 43%. Mass Spec (FD) 463. Calculated for C23 H15 ClF3 N3 OS: C, 58.29; H, 3.19; N, 8.87. Found: C, 58.56; H, 3.43; N. 9.02. M Pt 194°-195°C

PAC 1-[4-(2-Quinolylthio)phenyl]-3-(3,4-dichlorophenyl)thiourea

2-(4-Aminophenylthio)quinoline 11.9 mmoles, 3.0 g) and 3,4-dichlorophenylisothiocyanate 11.9 mmoles, 2.42 g) were reacted according to procedure B to yield the title compound, 360 mg, 6.6%. Mass Spec (FD) 455. Calculated for C22 H15 Cl2 N3 S2 : C, 57.90 H. 3.31; N, 9.21. Found: C, 57.86; H, 3.53; N, 9.07. m.p. 125°-126°.

The necessary amine starting material was prepared as follows:

a) 2-Chloroquinoline (0.12 moles, 20.0 g), 4-nitrothiophenol (0.13 mmoles, 20.8 g), and potassium carbonate (0.13 moles, 18.5 g) were dissolved in 800 ml ethanol and stirred at room temperature for 1.5 hr. The mixture was filtered and the solid washed with ethanol. The solid was dissolved with ethyl acetate, washed with water, dried over sodium sulfate and concentrated to yield 2-(4-nitrophenylthio)quinoline 26.0 g, 77%. Mass Spec (FD) 282. Calculated for C15 H10 N2 O2 S: C, 63.82, H 3.57; N, 9.92. Found: C, 63.55, H 3.59; N, 9.71.

b) 2-(4-Nitrophenylthio)quinoline (0.42 moles, 12.0 g) was hydrogenated in 300 ml DMF over 5.0 g Pd/C for 3 hr at room temperature. The solution was filtered through celite and concentrated. The product was triturated with ether and hexane to yield 2-(4-aminophenylthio)quinoline 10.0 g, 93%.

PAC 1-[4-(6-phenyl-3-pyridazylthio)phenyl]-3-(4-chloro-3-trifluoromethylphenyl) thiourea

3-(4-aminophenylthio)-6-phenylpyridazine (7.0 mmoles, 2.0 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (7.0 mmoles, 1.66 g) were reacted according to procedure C to yield of the title compound 2.5 g, 69%. Mass Spec (FD) 516. Calculated for C24 H16 ClF3 N4 O2 S2 : C, 55.76; H, 3.12; N, 10.84. Found: C, 55.98 H, 3.19; N, 10.78.

The necessary amine starting material was prepared as follows:

3-Chloro-6-phenylpyridazine (0.026 moles, 5.0 g), 4-aminothiophenol (0.026 moles, 3.3 g) and potassium carbonate (0.026 moles, 3.6 g) were stirred in 250 ml ethanol for 3 days. The reaction was filtered, concentrated, ethyl acetate added, washed with water and dried over sodium sulfate. The solution was concentrated and the product purified by HPLC over silica gel eluted with 50% ethyl acetate/hexane to yield 3-(4-aminophenylthio)-6-phenylpyridazine 6.0 g, 83%. Mass Spec (FD) 279. Calculated for C16 H13 N3 S: C, 68.79; H, 4.69; N, 15.04. Found: C, 68.87; H, 4.43N, 15.31.

PAC 1-[4-(2-pyridylthio)phenyl-3-(4-chloro-3-trifluoromethylphenyl)thiourea.

2-(4-Aminophenylthio)pyridine (6.0 mmoles, 1.2 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate (6.0 mmoles, 1.4 g) were reacted according to procedure C to yield the title compound, 1.9 g, 72%. Mass spec (FD) 439. Calculated for C19 H13 ClF3 N3 S2 : C, 51.88; H, 2.98, N, 9.55 Found: C, 51.99; H, 3.15; N, 9.63.

The necessary amine starting material was prepared as follows:

4-Aminothiophenol (0.065 moles, 8.1 g) in 275 ml DMF was stirred at room temperature for 1 hr with sodium hydride (0.065 moles previously washed with hexane). 2-Chloropyridine (0.06 moles, 6.8 g) was added and the reaction mixture was stirred for 6 hr. An additional 5.2 g of 4-aminothiophenol was added and the reaction stirred at room temperature for 16 hr. Water was added and the product extracted with ethyl acetate. The solution was washed thoroughly with water, dried over sodium sulfate and concentrated. The product was purified by HPLC over silica gel eluted with 30% ethyl acetate/hexane to yield 2-(4-aminophenylthio)pyridine 1.2 g, 10%. Mass spec (FD) 202. Calculated for C11 H10 N2 S: C, 65.32; H, 4.98, N, 13.85 Found: C, 65.23; H, 5.09; N, 13.63.

PAC 1-[4-(5-Nitroquinol-6-ylthio)phenyl-3-(4-chloro-3-trifluoromethylphenyl)thi ourea.

5-nitro-6-(4-aminophenylthio)quinoline 1.7 mmoles, 0.40 g) and 4-chloro-3-trifluoromethylphenylisothiocyanate 1.7 mmoles, 0.47 g) were reacted according to procedure C to yield the title compound, 0.78 g, 86% product. Mass spec (FD) 534. Calculated for C23 H14 ClF3 N4 O2 S2 : C, 51.64; H, 2.64, N, 10.47 Found: C, 51.84; H, 2.73; N, 10.43.

PAC 1-[2-(5-bromo-1,3,4-thiadiazolyl]-3-(2-naphthyl)urea

A solution of 2-naphthanoic acid(5.0 mmole, 0.86 g) and triethylamine (6.0 mmole, 0.61 g) in tetrahydrofuran(25 ml) cooled to 0° was treated with diphenylphosphoryl azide (5.5 mole, 1.51 g). The reaction was stirred at 22°C for 30 minutes, the reaction was treated with 2-amino-5-bromo-1,3,4-thiadiazole (5.0 mmole, 0.90 g). The reaction mixture was then stirred at 22°C for 30 minutes, then heated to 60°C After stirring at 60°C for approximately 22 hours, the reaction mixture was cooled to ambient temperature, the insolubles were collected by filtration and washed with tetrahydrofuran (2 ml). The filtrate was concentrated in vacuo to an oil. The oil was treated with methylene chloride (25 ml), washed with 0.1N sodium hydroxide (2×10 ml) hydrochloric acid (2×10 ml) and brine (10 ml). The organic phase was then subjected to silica gel column chromatography eluting with a 0 to 35% methanol:toluene. Fractions containing the desired material were combined and concentrated in vacuo to afford a solid. This material was treated with hot tetrahydrofuran (20 ml), filtered through Celite 521, reduced in volume and treated with hexane (6 ml). The insolubles were collected by filtration and washed with 3:1 hexane:tetrahydrofuran (9 ml). The filtrate was subjected to radial band chromatography to afford the title compound.

Analysis calculated for C13 H9 BrN4 OS: %C, 44.71; %H, 2.60; %N, 16.04. Found: %C, 44.97; %H, 2.77; %N, 15.83.

Field Desorption Mass Spectrum: M-1=348.

PAC 1-[2-(5-bromo-1,3,4-thiadiazolyl]-3-(1-naphthyl)urea

Following a method similar to that described in Example 23, but using 1-naphthylisocyanate instead of 2-amino-5-chloro-1,3,4-thiadiazole, the title compound was prepared.

Analysis calculated for C13 H9 BrN4 OS: %C, 44.71; %H, 2.60; %N, 16.04. Found: %C, 45.00; %H, 2.59; %N, 16.14.

Field Desorption Mass Spectrum: M-1=348.

Reel, Jon K., Simon, Richard L., Whitesitt, Celia A., Panetta, Jill A., Phillips, Michael L., Shadle, John K., Heinz, Lawrence J.

Patent Priority Assignee Title
10640457, Dec 10 2009 The Trustees of Columbia University in the City of New York Histone acetyltransferase activators and uses thereof
11034647, Dec 10 2009 The Trustees of Columbia University in the City of New York Histone acetyltransferase activators and uses thereof
11643394, Apr 30 2020 Icahn School of Medicine at Mount Sinai; THE UNITED STATES GOVERNMENT AS REPRESENTED BY THE DEPARTMENT OF VETERANS AFFAIRS; The Research Foundation for The State University of New York Krüppel-like factor 15 (KLF15) small molecule agonists in kidney disease
6583157, Jan 29 1998 Amgen Inc Quinolinyl and benzothiazolyl modulators
6653332, May 03 2000 Amgen Inc Combination therapeutic compositions and method of use
6727272, Jul 15 2002 Geneblue Corporation Leflunomide analogs for treating rheumatoid arthritis
6770648, Jun 30 1999 Amgen Inc Compounds for the modulation of PPARγ activity
7041691, Jun 30 1999 Amgen Inc Compounds for the modulation of PPARγ activity
7223761, Oct 03 2003 Amgen Inc Salts and polymorphs of a potent antidiabetic compound
7235576, Jan 12 2001 Bayer HealthCare LLC Omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
7329670, Dec 22 1997 Bayer HealthCare LLC Inhibition of RAF kinase using aryl and heteroaryl substituted heterocyclic ureas
7351834, Jan 13 1999 Bayer HealthCare LLC ω-Carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
7371763, Apr 20 2001 Bayer HealthCare LLC Inhibition of raf kinase using quinolyl, isoquinolyl or pyridyl ureas
7439242, Jan 29 1998 Amgen Inc PPARγ modulators
7517880, Dec 22 1997 Bayer HealthCare LLC Inhibition of p38 kinase using symmetrical and unsymmetrical diphenyl ureas
7528255, Feb 11 2002 Bayer HealthCare LLC Hydroxy, ω-carboxyaryl substituted diphenyl ureas and dirivatives thereof as raf kinase inhibitors
7601841, Jun 28 2000 Amgen Inc Quinolinyl and benzothiazolyl modulators
7625915, Dec 22 1997 Bayer HealthCare LLC Inhibition of RAF kinase using aryl and heteroaryl substituted heterocyclic ureas
7626033, Jun 30 1999 Amgen Inc Compounds for the modulation of PPARγ activity
7678811, Feb 11 2002 Bayer HealthCare LLC Pyridine, quinoline, and isoquinoline N-oxides as kinase inhibitors
7838541, Feb 11 2002 Bayer HealthCare LLC Aryl ureas with angiogenesis inhibiting activity
7897623, Jan 13 1999 Bayer HealthCare LLC ω-carboxyl aryl substituted diphenyl ureas as p38 kinase inhibitors
7928239, Jan 13 1999 Bayer HealthCare LLC Inhibition of RAF kinase using quinolyl, isoquinolyl or pyridyl ureas
7939551, May 03 2000 Amgen Inc.; Amgen Inc Combination therapeutic compositions
7960408, Jun 28 2000 Amgen Inc. Quinolinyl and benzothiazolyl modulators
7968567, Jun 30 1999 Amgen Inc. Compounds for the modulation of PPARγ activity
8003665, Oct 03 2003 Amgen Inc. Salts and polymorphs of a potent antidiabetic compound
8071616, Feb 11 2002 Bayer HealthCare LLC Pyridine, quinoline, and isoquinoline N-oxides as kinase inhibitors
8076488, Feb 28 2003 Bayer HealthCare LLC Bicyclic urea derivatives useful in the treatment of cancer and other disorders
8110587, Feb 11 2002 Bayer HealthCare LLC Aryl ureas as kinase inhibitors
8124630, Nov 28 2000 Bayer HealthCare LLC ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
8242147, Feb 11 2002 Bayer HealthCare LLC Aryl ureas with angiogenisis inhibiting activity
8618141, Feb 11 2002 Bayer HealthCare LLC Aryl ureas with angiogenesis inhibiting activity
8637553, Jul 23 2003 Bayer HealthCare LLC Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
8796250, May 20 2003 Bayer HealthCare LLC Diaryl ureas for diseases mediated by PDGFR
8841330, Nov 28 2000 Bayer HealthCare LLC Omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
9181188, Feb 11 2002 Bayer HealthCare LLC Aryl ureas as kinase inhibitors
9969677, Dec 22 2010 The Trustees of Columbia University in the City of New York Histone acetyltransferase modulators and uses thereof
Patent Priority Assignee Title
3990879, Dec 26 1974 Eli Lilly and Company Method of controlling aquatic weeds
4021225, Feb 26 1971 CHEVRON RESEARCH COMPANY, SAN FRANCISCO, CA A CORP OF DE Combating unwanted vegetation with 1,3,4-thiadiazole-2-sulfonamides
4174398, Mar 23 1977 Bayer Aktiengesellschaft Combating fungi with 1-alkyl-1-(1,3,4-thiadiazol-2-yl)-3-phenyl-ureas
4175081, Feb 01 1968 RHONE-POULENC, INC ; RHONE-POULENC, INC , A COP OF NY 5-Substituted thiadiazole ureas
4518414, Jan 22 1982 BASF Aktiengesellschaft Thiadiazole derivatives and their use for controlling undesirable plant growth
4576629, Mar 15 1984 RHONE POULENC NEDERLANDS B V Herbicidal thiadiazole ureas
4599424, Feb 01 1968 Rhone-Poulenc, Inc. 5-substituted thiadiazole ureas and their use as herbicides
4639526, Sep 19 1967 Bayer Aktiengesellschaft N-substituted 5-amino-1,3,4-thiadiazoles
4686294, Dec 20 1968 Ciba-Geigy Corporation 1,3,4-thiadiazolyl ureas and processes for controlling weeds and wild grasses therewith
4835168, Dec 16 1985 Eli Lilly and Company Thiadiazole antiviral agents
4876044, Oct 02 1969 Air Products and Chemicals, Inc Thiadiazole compounds and methods of use
4987233, Jun 15 1988 Eli Lilly and Company Process for preparing herbicidal ureas and insecticidal carbamates and carbamate derivatives
5087284, Feb 01 1968 Rhone-Poulenc, Inc. 5-substituted thiadiazole ureas and their use as herbicides
////////
Executed onAssignorAssigneeConveyanceFrameReelDoc
Mar 02 1995Eli Lilly and Company(assignment on the face of the patent)
Mar 14 1995HEINZ, LAWRENCE J Eli Lilly and CompanyASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0093130357 pdf
Mar 14 1995PANETTA, JILL A Eli Lilly and CompanyASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0093130357 pdf
Mar 14 1995PHILLIPS, MICHAEL L Eli Lilly and CompanyASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0093130357 pdf
Mar 14 1995REEL, JON K Eli Lilly and CompanyASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0093130357 pdf
Mar 14 1995SHADLE, JOHN K Eli Lilly and CompanyASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0093130357 pdf
Mar 14 1995SIMON, RICHARD L Eli Lilly and CompanyASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0093130357 pdf
Mar 14 1995WHITESITT, CELIA A Eli Lilly and CompanyASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0093130357 pdf
Date Maintenance Fee Events
Feb 26 2002M183: Payment of Maintenance Fee, 4th Year, Large Entity.
Apr 19 2006REM: Maintenance Fee Reminder Mailed.
Sep 29 2006EXP: Patent Expired for Failure to Pay Maintenance Fees.


Date Maintenance Schedule
Sep 29 20014 years fee payment window open
Mar 29 20026 months grace period start (w surcharge)
Sep 29 2002patent expiry (for year 4)
Sep 29 20042 years to revive unintentionally abandoned end. (for year 4)
Sep 29 20058 years fee payment window open
Mar 29 20066 months grace period start (w surcharge)
Sep 29 2006patent expiry (for year 8)
Sep 29 20082 years to revive unintentionally abandoned end. (for year 8)
Sep 29 200912 years fee payment window open
Mar 29 20106 months grace period start (w surcharge)
Sep 29 2010patent expiry (for year 12)
Sep 29 20122 years to revive unintentionally abandoned end. (for year 12)